Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Aug 19.
Published in final edited form as: Nat Immunol. 2022 May 27;23(6):848–860. doi: 10.1038/s41590-022-01224-z

Epigenetic regulation of T cell exhaustion

Julia A Belk 1,2,6, Bence Daniel 2,3,6, Ansuman T Satpathy 2,3,4,5
PMCID: PMC10439681  NIHMSID: NIHMS1924464  PMID: 35624210

Abstract

Chronic antigen stimulation during viral infections and cancer can lead to T cell exhaustion, which is characterized by reduced effector function and proliferation, and the expression of inhibitory immune checkpoint receptors. Recent studies have demonstrated that T cell exhaustion results in wholescale epigenetic remodeling that confers phenotypic stability to these cells and prevents T cell reinvigoration by checkpoint blockade. Here, we review foundational technologies to profile the epigenome at multiple scales, including mapping the locations of transcription factors and histone modifications, DNA methylation and three-dimensional genome conformation. We discuss how these technologies have elucidated the development and epigenetic regulation of exhausted T cells and functional implications across viral infection, cancer, autoimmunity and engineered T cell therapies. Finally, we cover emerging multi-omic and genome engineering technologies, current and upcoming opportunities to apply these to T cell exhaustion, and therapeutic opportunities for T cell engineering in the clinic.


T cells are a central component of the adaptive immune system and continuously survey the body for the presence of pathogens1. Each T cell is equipped with a somatically recombined T cell antigen receptor (TCR). Upon TCR recognition of foreign antigens presented on major histocompatibility complex (MHC) molecules, T cells are activated, clonally expand and kill infected cells using a variety of effector molecules. After clearance of infected cells, effector T cells differentiate into memory cells that persist long term and enable the rapid clearance of subsequent reinfections with the same pathogen. In contrast, in settings of chronic infection and cancer, foreign antigens often cannot be easily eliminated, and thus, T cells are chronically stimulated and can adopt a so-called exhausted cell state2. Functionally, exhaustion is characterized by low effector cytokine secretion, poor proliferative capacity and persistence, and the expression of inhibitory receptors on the cell surface, all of which can reduce the effectiveness of T cell-mediated immunity3. Although T cell exhaustion was initially described in the setting of chronic viral infections, it is now well appreciated that exhausted T cells (TEX) are present in many diseases, including in cancer, and that the gene regulatory programs governing exhaustion are largely conserved across disease settings4,5. Importantly, studies have suggested that T cell exhaustion limits the efficacy of immunotherapies, including checkpoint blockade and engineered cell therapies, and therefore, understanding the cellular and molecular regulation of TEX cells has become a central focus of the field69.

In this Review, we provide an in-depth discussion of the molecular regulation of T cell exhaustion with an emphasis on epigenetic mechanisms. We begin by discussing recent technologies that have enabled mapping epigenetic modifications at multiple scales in primary TEX cells, including histone modifications, transcription factors (TFs), chromatin accessibility, DNA methylation and genome conformation. We then describe how these methods have been leveraged to understand the epigenetic hallmarks of T cell exhaustion that have defined TEX cells as a distinct cell state, distinct cellular differentiation trajectories and regulatory pathways underlying the TEX cell fate, and lineage stability of TEX cells after antigen clearance or checkpoint blockade. Finally, we highlight emerging genome engineering and multi-omic technologies that may provide the next wave of insights into TEX cell programming and enable the development of new therapeutic strategies for cancer.

Technologies to interrogate epigenetic regulation

In mammalian cells, epigenetic mechanisms enable cells to differentiate, adapt to changes in the environment and propagate their cellular state after cell division10. Each chromosome is organized into megabase-sized topologically associated domains (TADs), which are largely conserved from early stem cells to differentiated cell types. Within this invariant genome scaffold, cell-type-specific and cell-state-specific gene regulatory DNA interactions establish specific gene expression programs, such as T cell exhaustion. This bridge from identical genotype (DNA sequence) to disparate phenotypes (gene expression) is broadly termed ‘epigenetic regulation’ and is coordinated by a complex interplay of cis-acting DNA elements (enhancers) and trans-acting TFs. The development and application of genome-scale technologies to systematically map features of the epigenome has uncovered principles of TEX cell genomic organization at multiple scales, including histone modifications and TFs, chromatin accessibility, DNA modifications such as DNA methylation and three-dimensional (3D) folding of the genome.

Fundamental DNA elements required for efficient gene transcription are the promoter, transcription start site (TSS) and distant gene regulatory enhancers11 (Fig. 1a). The promoter is upstream of the gene TSS and provides a platform for the assembly of the general transcription factor (GTF) complex, which is necessary for RNA polymerase II (RNAPII) recruitment and gene transcription12. Enhancers are non-protein-coding DNA elements that may be far away from the gene in the linear genome, but can be bound by cell-type-specific TFs and brought into spatial proximity of the gene TSS by genome conformational changes to activate gene transcription with RNAPII13. Dense clusters of enhancers, termed ‘super-enhancers,’ are defined by high occupancy of the Mediator complex and master TFs1416. Super-enhancers preferentially regulate key lineage-determining TFs and signaling molecules, such as cytokines and cytokine receptors in T cells16,17.

Fig. 1 |. Epigenetic regulation of gene expression.

Fig. 1 |

a, Promoters, enhancers and insulators are shown in their active and repressed states. b, Technologies to map protein localization on chromatin (ChIP–seq), accessible chromatin (ATAC–seq) and DnA methylation (bisulfite-seq). c, Technologies to map spatial genome architecture. eRnA, enhancer RnA; GTF, general transcription factor; MBD, methyl-CpG binding domain; P-TeFb, positive transcription elongation factor.

Central to the spatial organization of the genome is chromatin, the complex of DNA and proteins that structures the genome and packages it into the nucleus18. To fit nearly 2 meters of DNA into the ~10-μm nucleus of each single cell, DNA is first packaged into nucleosomes, which consist of ~147 base pairs of DNA wrapped around a histone octamer, and then into larger and larger chromatin fibers that form chromosomes19. Chemical modifications of chromatin regulate gene transcription20. Post-translational modifications (PTMs) of histone tails can either repress or activate gene transcription (Fig. 1a). For example, lysine acetylation of histone tails, such as acetylated histone H3 Lys27 (H3K27ac) and acetylated histone H3 Lys9 (H3K9ac), is associated with active promoters and enhancers and is deposited by histone acetyltransferases10. Lysine acetylation activates transcription via recognition by bromodomain-containing proteins (for example, BRD4), which in turn facilitate transcriptional elongation by interacting with the positive transcription elongation factor complex that phosphorylates the C-terminal domain of RNAPII21,22. Conversely, histone deacetylases remove this modification and thereby repress gene expression. Similarly, lysine methylation can represent either repressed (for example, H3K9me3 and H3K27me3) or active (for example, trimethylated histone H3 Lys4 (H3K4me3)) chromatin states and is deposited by histone methyltransferases, such as EZH2, which deposits H3K27me3. Histone methylation can be reversed by histone lysine demethylases (Fig. 1a). Finally, bivalent gene loci are those that are simultaneously marked by activating (for example, H3K4me3) and repressive (for example, H3K27me3) modifications, which signify a ‘poised’ chromatin state that enables rapid control of gene expression23. For example, in naïve CD8+ T cells, gene loci encoding TFs important for differentiation, including the Tbx21, Irf4, Gata3 and Eomes loci, are maintained in a bivalent state that enables rapid cell fate commitment upon TCR signaling24.

The location of specific histone modifications and TFs can be mapped genome wide with methods that link antibody-mediated immunoprecipitation or immunocleavage of proteins or PTMs with high-throughput sequencing. One technology developed for this purpose is chromatin immunoprecipitation with sequencing (ChIP–seq), where genomic DNA is crosslinked to associated proteins or PTMs and then fragmented and immunoprecipitated. Sequencing of the DNA pulled down with the target of interest reveals the genome-wide localization of the given protein or histone modification2528 (Fig. 1b). Recent adaptations of this method use antibody-tethered micrococcal nuclease (MNase) or Tn5 complexes to cut chromatin at sites proximal to a bound protein, termed chromatin immunocleavage (ChIC)29. These cleaved DNA fragments then diffuse out of the nucleus and can be isolated and sequenced. When MNase is used, the method is known as CUT&RUN (or ChIC-seq), and when Tn5 is used, the method is known as CUT&TAG (or ACT-seq)3033. Each of these methods obtain similar information to ChIP–seq without the requirement for cellular fixation. Further, they often reduce the required input cell number, enabling profiling of primary cell types3436 and even single cells, as recently demonstrated with single-cell CUT&TAG and single-cell ChIC-seq3033,37,38. It is important to note that inefficiencies in immunocleavage and signal dropout inherent to sequencing technologies can limit the insights gained from single-cell adaptations of these methods, for example, specific TF-binding sites in each cell, and therefore, aggregation of signals from many single-cell profiles may be required39.

A fundamental measurement of chromatin structure is accessibility; namely, is a regulatory DNA sequence open (and active) or closed (and inactive)? Inactive genes and regulatory elements are generally organized into densely packed chromatin fibers, called heterochromatin, whereas transcribed genes and active regulatory elements typically remain in loosely packaged euchromatin. Furthermore, some stretches of DNA within euchromatin are depleted of nucleosomes and can directly interact with TFs or other transcriptional machinery to control gene expression. The outcome of this genome structure is that only a small fraction of the genome—approximately 1–2%—is accessible at any given time in a cell, and the identification of these open sites can be used to nominate sites that may regulate gene expression40.

The location of accessible DNA genome wide can be profiled with technologies such as DNase-seq, MNase-seq and assay for transposase-accessible chromatin with sequencing (ATAC–seq)4144. The common principle underlying each method is the enzymatic cleavage of chromatin, either by DNase-I, MNase or Tn5. These enzymes preferentially cut open chromatin, and therefore, sequencing the resulting DNA fragments identifies genomic locations that are accessible. Cis-regulatory sites can be identified by enrichments of reads (for example, representing DNase-I hypersensitive sites in DNase-seq) at specific regions of the genome. The activity of trans-factors can be inferred through the identification TF ‘footprints’ within these sites—DNA sequences that are protected from enzymatic cleavage via direct binding of a TF. Thus, the locations of thousands of potential cis- and trans-elements can be read out genome wide using a single assay. For example, ATAC–seq utilizes a hyperactive prokaryotic transposase (Tn5) to directly transpose sequencing adaptors into regions of accessible chromatin44. As with DNase-seq, ATAC–seq profiles can provide insights into several layers of epigenetic regulation from a single assay, including the identification of enhancer and promoter sequences genome wide with base-pair resolution, the precise positioning of nucleosomes, and the inference of TF activity through DNA footprinting of transposase-inaccessible regions4446. However, it is important to note that while TF footprint predictions are often accurate in aggregate (that is, when averaged across the genome), they typically cannot conclusively identify specific TFs that are active at individual DNA positions and require experimental validation to demonstrate a functional role for the TF. Initially, the major advantages of performing ATAC–seq compared to DNase-seq were its lower preparation time (several hours compared to days) and associated cost, and its sensitivity for samples with low cell numbers, or for archival tissues or sections44,47. In recent studies, DNase-seq has also been adapted for low-input and archival samples, even down to single-cell resolution48,49. Overall, these technological advances in open chromatin analysis have enabled epigenetic profiling of mouse and human TEX cells, including intratumoral T cells in clinical samples, discussed below68,50.

Another important regulator of gene expression involves DNA methylation that occurs upon covalent addition of a methyl group to the pyrimidine ring of cytosine to form 5-methylcytosine (5-mC)51. The presence of 5-mC on promoters and enhancers typically indicates repression of gene transcription but has also been reported to facilitate transcription52. At repressed genomic regions, 5-mC is bound by methyl-CpG binding proteins, which facilitate chromatin condensation (Fig. 1a). DNA methylation is catalyzed by DNA methyltransferases (DNMTs) and is oxidized by TET enzymes into 5-hydroxymethyl cytosine (5-hmC), which can then be converted back to unmodified cytosine via multiple mechanisms53. Among DNMTs, DNMT3 enzymes (DNMT3A and DNMT3B) mediate de novo methylation, while DNMT1 maintains the methylation landscape during cell division54. The location of methylation genome wide can be mapped by bisulfite sequencing (bisulfite-seq; Fig. 1b). Bisulfite treatment of DNA chemically converts cytosine to uracil, but cytosines marked by 5-mC or 5-hmC are protected. Therefore, identities of methylated and unmethylated cytosines can be distinguished by sequencing5557.

The 3D conformation of chromosomes underlies the spatial interaction of distal regulatory elements with target genes, and recent advances in sequencing-based technologies have improved our understanding of the multi-scale organization of the 3D genome13,58 (Fig. 1a). These methods typically begin by crosslinking cells, which preserves interactions between neighboring and distant genomic loci. Next, cells are lysed, and DNA is digested with a restriction enzyme followed by proximity ligation to generate small fragments of interacting DNA. The fragments of interest are chimeric; they contain fragments of DNA from two interacting locations in the genome, and these ‘contacts’ are identified as chimeric reads from sequencing (Fig. 1c). Chromosome conformation capture (Hi-C) was the first technology that enabled the unbiased and systematic mapping of all genome-wide DNA contacts58,59. Initial Hi-C maps demonstrated that chromosomes can be characterized by spatially separated compartments that can be several megabases in size. Within compartments, TADs represent the next level of genome organization and are self-interacting loci in the range of 0.2–1.0 Mb60,61. The main architectural regulators of genome conformation are insulator elements, which bind TF CTCF at TAD boundaries. TADs form via a dynamic process of cohesin-mediated loop extrusion62, which can proceed either symmetrically (that is, both cohesin rings slide equally along DNA) or asymmetrically, as occurs when one cohesin ring is stuck at a particular location and ‘reels in’ the other ring63. Asymmetric extrusion can be caused by tethering of promoters within the loop to super-enhancers, manifesting as a ‘stripe’ on the Hi-C contact map and termed a ‘stripe domain’. Recent adaptations of Hi-C have improved its sensitivity (for example, in situ Hi-C) and have also incorporated a protein-directed immunoprecipitation step (for example, HiChIP)6467 (Fig. 1c). These more sensitive technologies have revealed sub-TAD structures, such as specific enhancer–promoter DNA loops, as well as structures that are bound by proteins or chemical modifications of histones. Importantly, newer technologies, including HiChIP, have dramatically reduced the cell number required for profiling protein-directed genome conformation, enabling the application of these technologies to TEX cells, as recently demonstrated in chimeric antigen receptor (CAR) T cells68. Altogether, epigenetic modifications, chromatin accessibility and genome conformation provide spatiotemporal control of gene regulation, and the recent development of these genomic technologies with increased sensitivity, resolution and throughput have enabled new insights into the molecular mechanisms underpinning T cell function.

Distinct epigenetic programs in TEX cells

The central function of T cells is to monitor peptide antigens presented by MHC molecules, discriminate self from foreign antigens and, upon recognition of a foreign antigen, mount an appropriate immune response1. The T cell response is directed not only by the TCR–antigen signal (signal 1), but also by co-stimulatory signals (signal 2) and the local cytokine milieu (signal 3)69. Collectively, these signals induce the activation and differentiation of naïve T cells into different T cell subsets, each with distinct phenotypes and functions. After activation, naïve CD8+ T cells differentiate into a highly proliferative and cytotoxic effector state70 (Fig. 2a). If the antigen can be cleared, such as in acute viral infections, most effector cells undergo apoptosis (termed short-lived effector cells or terminal effector (TE) cells), while a small fraction persist and differentiate into memory cells via memory precursor (MP) cells71. MP cells and TE cells can be distinguished via reciprocal expression of surface markers KLRG1 and interleukin (IL)-7R, and the fate decision is established by the TF T-bet; high Tbx21 (which encodes T-bet) expression induces the TE cell fate, while low expression induces the MP cell fate71,72. Early epigenetic profiling studies in primary CD8+ T cells demonstrated how DNA methylation, histone modifications and chromatin accessibility regulated the processes of T cell activation, effector response and memory formation during viral infection24,73,74. For example, these studies demonstrated that key effector genes, such as Prf1 and Gzmb, are demethylated and gain chromatin accessibility upon antigen recognition, while naïve-associated genes are repressed. After pathogen clearance, MP cells can differentiate into long-lived memory cells by demethylating naïve-associated genes required for survival, such as Bcl2 and Il7r. As a result, memory CD8+ T cells have demethylated and open chromatin at both effector and naïve genes, which enable them to be long-lived, while retaining the ability to rapidly mount an effector response upon pathogen reencounter73,74.

Fig. 2 |. CD8+ T cell development and key mediators of T cell exhaustion.

Fig. 2 |

a, Differentiation trajectory of CD8+ T cells. Important surface markers and TFs are indicated on each cell type. b, Key transcriptional regulators of CD8+ T cells and exhausted CD8+ T cells. IFn-γ, interferon gamma; TnF, tumor necrosis factor.

In contrast to acute infection, in settings of persistent antigen, such as in chronic viral infections or cancer, CD8+ T cells can instead become exhausted. T cell exhaustion is characterized by poor proliferative potential, expression of multiple cell surface inhibitory receptors (for example, PD-1, CTLA-4, TIM3 and LAG3) and a loss of effector function2. Although TEX cells were initially characterized in the context of the chronic murine lymphocytic choriomeningitis virus (LCMV) clone-13 infection model, there is a growing appreciation that these cells are conserved across a variety of disease settings, including human infectious diseases, cancer and autoimmune disease7579, and are a key determinant of response to immunotherapies, including checkpoint blockade and engineered T cell therapies5,8083. However, whether T cell exhaustion represents a distinct cell state and differentiation program—as opposed to the isolated upregulation of a select few inhibitory receptors, such as PD-1—remained unresolved until recently8486. To address this question, multiple studies profiled LCMV-specific or tumor-specific CD8+ T cells to identify open chromatin sites associated with T cell exhaustion69,87. In both models, comparative analysis between TEX cells and naïve, effector or memory T cells, or between tumor-infiltrating TEX cells and acutely stimulated or bystander T cells, revealed genome-wide reprogramming of the TEX cell epigenetic state, comprising thousands of differentially accessible regions, many of which neighbored genes that mediate TEX cell differentiation. For example, TEX cells lacked open chromatin sites present in the Ifng locus in T effector and memory cells, which accompanied the diminished expression of Ifng in TEX cells. Similarly, several TEX cell-specific open chromatin sites were present in the Pdcd1 locus, and CRISPR–Cas9 mutagenesis experiments demonstrated that these sites functioned as bona fide enhancers that maintain high levels of PD-1 expression specifically in TEX cells6. These findings, taken together with previous studies demonstrating large-scale transcriptional changes in TEX cells86, support the concept that TEX cells represent a distinct T cell chromatin state, rather than the isolated expression of inhibitory receptors.

Importantly, epigenetic analysis in human chronic infection, such as human immunodeficiency virus (HIV), and in human cancers, has demonstrated a conserved epigenetic profile between murine and human TEX cells4,6,50,75. ATAC–seq profiling of TEX cells in the setting of human basal cell carcinoma identified ~4,500 differentially accessible regions in TEX cells50. This extent of chromatin remodeling was comparable to that which was observed in other T cell states, such as regulatory CD4+ T cells or effector CD8+ T cells, indicating a large-scale change that is consistent with a distinct cell lineage, as was observed in murine TEX cells. Accordingly, a core TEX cell gene signature derived from transcriptomic and epigenomic profiling of murine TEX cells in chronic LCMV infection was shared in human TEX cells present in chronic infection and cancer4. More broadly, a recent study performed a comprehensive reanalysis of over 300 human and mouse ATAC–seq and RNA-sequencing (RNA-seq) datasets from CD8+ T cells in chronic infection and cancer and showed that T cells obtained from both settings exhibited highly similar global chromatin profiles, although precise enhancer sequences in individual gene loci may diverge across organisms68,75. Altogether, these results demonstrate that TEX cells exhibit a common differentiation program across species and immune challenges, which suggests that this program may be regulated by common upstream signals and TEX cell-specific TFs.

Subsets of exhausted T cells and key transcription factors

Temporal and TEX cell subset-based analysis has further enabled the dissection of epigenetic and TF programs underlying TEX cell differentiation. A temporal analysis of tumor-infiltrating lymphocyte (TIL) exhaustion over the course of 60 days after the transfer of naïve T cells into the tumor microenvironment (TME) identified two distinct phases of chromatin remodeling8. The first phase of remodeling occurred early, within 5 days of T cell transfer, while the second (and final) phase of remodeling occurred approximately 2 weeks after transfer. The number of differentially regulated cis-regulatory elements was similar in both phases, but these sites were regulated by different TFs. Comparing chromatin changes in early (day 7) versus late (day 14) TILs revealed that day 7 TILs had increased chromatin accessibility at sites containing AP-1, NFAT and TCF-1 TF motifs, while day 14 TILs had increased accessibility at sites containing E2F and KLF TF motifs. To test the functional importance of these regulatory programs, TILs were isolated at multiple time points for functional studies, which demonstrated that early TEX cells (day 5, PD-1hiCD38loCD101lo cells) could regain effector function when removed from the TME, while late TEX cells (day 12 or after, PD-1hiCD38hiCD101hi) could not, leading to a model in which PD-1lo TILs undergo two sequential waves of chromatin remodeling, of which only the early TEX cell epigenetic program may be reversible. Interestingly, transplanting memory T cells into tumor-bearing mice revealed that the TME could induce a similar epigenetic state in naïve and memory T cells.

These results suggested a stepwise acquisition of the epigenetic program of exhaustion, and the early epigenetic changes suggest a distinct cellular TEX cell differentiation trajectory after T cell activation, compared to effector and memory T cells. Indeed, flow cytometry analysis of antigen-specific T cells in chronic LCMV infection identified the presence of a progenitor TEX cell population that developed early during chronic infection, which was defined by its ability to proliferate and self-renew in response to antigen, its preferential localization in lymphoid organs, and the expression of the TF TCF-1 (refs. 5,80,88). Moreover, after anti-PD-1 immunotherapy, these cells were the primary source of the T cell proliferative burst, suggesting their preserved function, compared to other TEX cell populations80. In subsequent studies, two additional subsets of TEX cells have been described: transitory TEX cells, defined by their expression of PD-1 and CX3CR1, and terminal TEX cells, defined by their expression of PD-1, TIM3, LAG3, CD38, CD39 and CD101 (refs. 8890) (Fig. 2a). Transitory TEX cells exhibit substantial proliferative and effector function and represent an intermediate cell state between progenitor TEX and terminal TEX cells, which exhibited the most severe functional defects. Importantly, although studies have demonstrated that terminal TEX cells develop from transitory TEX cells88,91, under certain circumstances—including insufficient CD4+ T cell help89 or the presence of transforming growth factor-β (TGF-β)92— it has also been proposed that cells may progress directly from progenitor TEX cells to terminal TEX cells. In these cases, CX3CR1+ cells have been proposed to represent an alternate endpoint that does not progress to terminal TEX cells89,92. Additional lineage tracing studies will be needed to determine whether the CX3CR1+ state is truly bypassed in these situations or the differentiation to terminal TEX cells is simply accelerated.

TCR-based lineage tracing analysis of antigen-specific T cells in chronic LCMV infection supports the concept that a subpopulation of transitory TEX cells may represent an alternate TEX cell differentiation endpoint93. This study identified heterogeneity within the CX3CR1+ transitory TEX cell pool and proposes a model in which TEX cells progress through a CX3CR1+ TEX cell intermediate phenotype to either terminal TEX cells or an alternate CX3CR1+ TEX cell state, which is marked by expression of killer cell lectin-like receptors (KLRs; TEXKLR)93. Therefore, three non-mutually exclusive models of differentiation are emerging: (1) a linear model in which all cells eventually progress to terminal TEX cells (Fig. 2a), (2) a linear model in which certain cells do not progress past transitory TEX cells (for example, certain cells are retained in a CX3CR1+ TEXKLR state) and (3) a divergent model in which cells progress from progenitor TEX cells to either terminal TEX cells or transitory TEX cells (including TEXKLR). The determinants of TEX cell differentiation trajectories may depend on multiple factors. As one example, an important determinant of T cell fate is TCR affinity, which manifests via TCR signal strength94. It has been shown that when two antigens of different affinities are present within the same tumor, T cells specific for the weaker antigen are enriched for progenitor TEX cells95. In addition, recent complementary studies analyzing polyclonal T cells responding to a single antigen (gp33)93 or transgenic T cells responding to three antigens of carefully defined affinities94, have demonstrated that TCRs with higher affinity may preferentially progress to terminal TEX cells, while lower-affinity interactions induce alternate states including TEXKLR cells. In CAR T cells, repeated antigen exposure has also been shown to induce a natural killer cell-like state, which expresses many of the same genes as TEXKLR cells96.

To further understand the regulatory programs underlying these functional state transitions, recent studies have performed ATAC–seq analysis of TEX cell subsets in the setting of chronic LCMV infection91,97,98. Analysis of TF motif accessibility across subsets identified early-stage and late-stage TF activities, analogous to the observations in exhausted TILs. Progenitor TEX cells showed increased activity of TCF-1 and BACH2, transitory TEX cells showed enrichments in T-bet and RUNX motifs, and terminal TEX cells showed enrichments in NR4A and EOMES motifs, nominating a hierarchy of TF families whose activity may underlie each cell-state transition. Importantly, although transitory TEX cells expressed several transcripts and TFs in common with effector T cells (for example, Cx3cr1, Tbx21, and so on), these cells were epigenetically distinct, with nearly 5,000 differentially accessible regulatory elements compared to effector cells. Similarly, H3K27ac ChIP–seq analysis demonstrated that progenitor TEX cells exhibited the most distinct active enhancer landscape (2,863 unique enhancers) compared to terminal TEX, MP and TE cells in LCMV infection99. A common finding across multiple modalities, including ATAC–seq, ChIP–seq and HiChIP, is the enrichment of AP-1/bZIP family TF motifs in active chromatin of terminal TEX cells36,68,83, which may represent promising targets for functional follow-up and engineering. Interestingly, comprehensive CUT&RUN analysis of active and repressed chromatin marks in exhausted TILs revealed a decoupling of active histone modifications and active gene expression, as well as an increase in bivalent enhancers, suggesting the presence of altered 3D chromosome conformation in terminal exhaustion36. Accordingly, H3K27ac HiChIP in naïve, exhausted (HA-28z) and non-exhausted CAR T (CD19–28z) cells identified TEX cell-specific 3D chromatin conformation, which in many cases exhibited differential chromatin looping despite minimal changes in chromatin accessibility, suggesting a further layer of TEX cell genome regulation that should be investigated in future studies68. Altogether, these findings support a distinct lineage trajectory for TEX cells and nominate sequential TF activities that may program the TEX cell epigenetic state.

Despite these advances, the existence and identity of a lineage-determining TEX cell TF remained unknown. By analyzing differentially expressed genes in TEX cells across chronic infection and mouse and human tumors, several studies identified the TF TOX as a key regulator of the TEX cell lineage99103. In chronic infection and in tumors, TOX was rapidly induced by TCR signaling in TEX cells and remained highly expressed, while in acute infections, low levels of TOX were transiently induced but not sustained101,102. Importantly, overexpression of TOX in T cells in vitro was sufficient to recapitulate several features of the TEX cell program, including upregulation of the inhibitory receptor genes, Pdcd1 and Havcr2, and Entpd1; however, the magnitude of gene expression change was not as large as is observed in TEX cells in vivo. Conversely, genetic deletion of Tox led to a decrease in the surface expression of inhibitory receptors and improved proliferation in tumors and during chronic infection but did not impact the development of effector or memory cells during acute infection. Intriguingly, not all aspects of T cell exhaustion were reversed by the deletion of Tox; namely, Tox-deficient and wild-type T cells showed varied defects in the production of effector molecules and in their ability to lyse antigen-bearing cells. It remains to be determined whether these discrepancies are due to the different model systems tested or other factors. Moreover, Tox-deficient cells persisted less in tumors, compared to wild-type cells, suggesting a decoupling of multiple TEX cell programs (for example, inhibitory receptor expression, effector functions and persistence) downstream of TCR signaling. These data suggest that TOX may work in concert with other key TFs to establish the full TEX cell program.

At the epigenetic level, transcriptional changes observed in Tox-deficient cells, or after Tox overexpression, were associated with corresponding changes in chromatin accessibility101103. For example, putative regulatory elements in the Pdcd1, Cd38 and Entpd1 gene loci were less accessible in Tox-deficient T cells, compared to wild-type T cells. Globally, approximately 40% of the accessible regions that were significantly decreased in Tox-deficient cells were TEX cell specific. In contrast, epigenetic analysis revealed an increase in accessibility in a large fraction of sites near effector genes, including Klrg1, Gzma, Gzmb and Zeb2, supporting a role for TOX as a key determinant of the early fate decision between effector and TEX cell lineages. ChIP–seq analysis revealed that the Tox locus was bound by NFAT1 and NFAT2, key TFs immediately downstream of TCR signaling, and chronic TCR stimulation and NFAT2 overexpression were sufficient to induce Tox expression, while NFAT2-deficient T cells failed to upregulate Tox101. However, the sustained expression of Tox appears to be independent of NFAT2 and may be regulated at least in part by DNA methylation of the Tox locus100.

Taken together with previous epigenetic studies, these results suggest a temporally coordinated TF hierarchy that establishes and maintains the TEX cell state (Fig. 2b). First, immediately downstream of TCR signaling, NFAT proteins tune the balance between productive T cell activation and T cell dysfunction104. NFAT:AP-1 heterodimers lead to T cell activation, while ‘partnerless’ NFAT directly binds to and induces expression of inhibitory receptor genes and Tox102,104. Progenitor TEX cells maintain expression of Tcf7 (which encodes TCF-1 and is also expressed in naïve T cells), which may initially be driven by BACH2 and enables them to self-renew and proliferate98. However, following continued antigen stimulation, T cells proceed to a transitory TEX cell state, which has partial effector activity, driven by the activity of T-bet. Finally, T cells progress to terminal TEX cells, in which sustained expression of TOX induces the upregulation of EOMES and NR4A TFs. These factors in turn regulate the terminal exhaustion program that includes inhibitory receptors, decreased proliferative and effector functions, and increased pro-survival molecules and metabolic adaptations that ensure T cell persistence in the setting of chronic antigen.

Epigenetic stability of T cell exhaustion

The clinical success of immunotherapies targeting inhibitory surface receptors on T cells, including PD-1 and CTLA-4 blockade, has motivated research into the T cell subsets responsible for tumor control and clinical response80,81. T cell infiltration and exhaustion have been associated with clinical response, but it has been unclear whether TEX cells are a cause or a byproduct of tumor regression. Early studies on the effect of PD-1 blockade on CD8+ T cells were performed in the setting of chronic viral infection, where it was shown that treatment could lead to the expansion of highly functional antigen-specific CD8+ T cells, in addition to improved survival and reduced viral load76,77,105,106. Adoptive transfer of congenically marked TEX cells induced by chronic infection showed that these cells maintain substantial proliferative capacity, leading to a model in which reinvigoration of preexisting TEX cells directly mediates disease response. In contrast, other functional studies of TEX cells after adoptive transfer demonstrated persistent impairments in effector function and cytotoxicity even after the removal of antigen107.

Recent studies have revisited this concept with genome-wide transcriptional and epigenetic profiling technologies and from the epigenetic viewpoint of TEX cells. The precise definition of the TEX cell-specific chromatin signature allowed one group to investigate the stability of this chromatin state in the setting of immunotherapy7. Strikingly, after PD-1 blockade, only ~10% of the epigenetic landscape was ‘reinvigorated’ to resemble the effector T cell landscape, suggesting that the durable reacquisition of T cell function may be limited by the stability of the TEX cell-associated chromatin state. In addition, a small subset of chromatin regions (98) diverged even further from the effector state after anti-PD-L1 treatment. The 555 chromatin regions that demonstrated at least partial reversal to the effector state were enriched for NFAT-binding sites, and ‘partnerless’ NFAT-dependent genes showed reduced expression in anti-PD-L1-treated TEX cells, highlighting that several coordinated TF pathways may underlie TEX cell stability7,104.

A second series of studies took this question a step further and asked whether the TEX cell chromatin state remains stable even in the absence of antigen108110. In one of these studies, the authors transferred TEX cells from chronic LCMV-infected mice to infection-free mice and analyzed their functional, transcriptional and epigenetic reinvigoration toward T effector cells108. Strikingly, even though TEX cells acquired some transcriptional features of the memory T cell program, such as the downregulation of inhibitory receptors and reexpression of Il7r and Tcf7, these cells were still highly impaired in their ability to proliferate and generate a robust recall response in the context of a new infection. Importantly, ATAC–seq analysis showed that the chromatin state of TEX cells transferred into an infection-free animal still more closely resembled the TEX cell state, rather than the T cell memory state; namely, only 182 regulatory elements changed accessibility after removal of antigen. These results suggest that the TEX cell epigenetic state is highly stable, indicating that chronic antigen exposure leaves persistent ‘scars’ that are not removed by PD-1 blockade or cessation of antigen exposure. Finally, the authors determined that TEX cells that could proliferate in the recall response were predominantly derived from progenitor TEX cells, supporting the relative reversibility of early TEX cell programs, compared to late TEX cell programs.

Although the precise molecular programs that maintain the TEX cell state are still under investigation, one study has demonstrated that this is mediated at least in part by the de novo DNA methyltransferase, DNMT3A111. Whole-genome bisulfite-seq of antigen-specific CD8+ T cells in LCMV clone-13-infected mice identified ~1,200 DNA methylation events that accompanied the TEX cell transition. Analysis of Dnmt3a-conditional knockout mice demonstrated that these methylation events were DNMT3A-dependent and included target genes such as Eomes, Tbx21 and Tcf7 (ref. 111). Interestingly, this de novo exhaustion methylation program was not impacted by PD-L1 blockade, but anti-PD-L1 blockade synergized with DNMT3A inhibition to enhance T cell proliferation in chronic infection. Of note, DNMT3A may also represent a promising target for CAR T cell engineering, as DNMT3A-knockout CAR T cells demonstrated enhanced antitumor activity, proliferation and effector function while limiting exhaustion112.

In summary, these epigenetic studies support several fundamental concepts regarding the regulation of T cell exhaustion. First, that the primary driver of TEX cell lineage commitment is chronic TCR signaling, not environment-specific effects or PD-1 signaling. This concept is supported by the conservation of a common TEX cell program across diverse disease settings with different microenvironment effects4,5,75, and in CAR T cells83, the induction of Tox expression by chronic TCR signaling or downstream TFs102,104, the similarity of exhaustion programs in CD8+ and CD4+ T cells that experience chronic TCR signaling50,113, comparisons of tumor-specific TILs with bystander TILs in the same tumor environment9,114, and comparisons of antigen-specific T cells responding to variants of chronic LCMV infection that allow the strength and duration of antigen stimulation to be varied in a controlled manner100,115. Second, that in many cases, T cell exhaustion may be beneficial for the organism, enabling T cells to continue to persist in the setting of chronic antigen stimulation with reduced function, rather than undergoing activation-induced cell death. Therefore, targeting select, but not all, molecular programs in TEX cells may provide a ‘goldilocks’ approach for improved T cell function in cancer94.

Targeted perturbation of the epigenome and single-cell technologies

New technologies for targeted genetic and epigenetic perturbations, as well as single-cell and multi-omic profiling methods, are an important frontier for characterizing immune cell function and T cell exhaustion. CRISPR–Cas9-based genome engineering has made genetic and epigenetic perturbations easier, faster and higher fidelity than was previously possible116,117. There are two versions of CRISPR–Cas9 that provide the basis for an extensive suite of emerging technologies (Fig. 3a). The first, and original function of Cas9, is to edit the genome at a location programmed by a single guide RNA (sgRNA; Fig. 3a). In most cases, the induced double-strand break is repaired by nonhomologous end joining, which results in a small random insertion or deletion (indel) at the cut site. These indels are typically deleterious and, when targeted to coding regions of the genome, result in disruption of the targeted protein. Alternatively, if an exogenous piece of template DNA is provided with ends homologous to the sequences flanking the cut site, the template may be integrated into the genome via homology-directed repair. A second adaptation of CRISPR–Cas9 is catalytically dead Cas9 (dCas9), which retains the genome targeting capability of wild-type Cas9 but is not able to induce double-stranded breaks118 (Fig. 3a). Therefore, any protein that can be expressed and function as a fusion construct with dCas9 can be precisely targeted to any genomic location, which is useful for many applications, including transcriptional inhibition (CRISPRi)118,119 or activation (CRISPRa)120. Finally, both Cas9 and dCas9 can be used together with pools of sgRNAs (as opposed to individual sgRNAs) to perform high-throughput screens of coding120123 and noncoding regions124126. Although initial proof-of-concept studies were performed in model systems (for example, cell lines), the development of the Cas9-expressing mice and methods to efficiently deliver Cas9 into primary T cells are enabling broader applications of genome engineering technologies to understand immune regulation35,126134. In the context of T cell exhaustion, recent studies have demonstrated the ability of putative enhancers identified by epigenetic studies to be functionally tested with CRISPR, for example, validating a causal role for several cis-regulatory elements in the regulation of PD-1 expression6,68. Pooled screening in TEX cells has also enabled functional interrogation of TFs and led to the identification of Fli1 as a factor that limits T cell function35. We envision that future studies will soon enable the large-scale testing of putative regulatory elements and TFs nominated by epigenetic profiling to rapidly uncover functional elements and novel TEX cell biology.

Fig. 3 |. Emerging technologies for studying the epigenome.

Fig. 3 |

a, CRISPR–Cas9-based technologies for perturbing the epigenome. b, Multi-omic technologies for cell profiling. HDR, homology-directed repair; nHeJ, nonhomologous end joining.

In parallel, single-cell technologies for profiling the transcriptome135, epigenome50, immune receptor repertoire136, surface proteins137,138 and CRISPR perturbation139 have matured into streamlined and widely available platforms (Fig. 3b). There have also been substantial advances in combining multiple modalities within the same cell, for example, in CITE-seq (single-cell RNA-seq with surface proteins)138, SHARE-seq (single-cell RNA-seq with single-cell ATAC–seq)140, Perturb-seq (single-cell RNA-seq with CRISPR)141143, Perturb-ATAC (single-cell ATAC–seq with CRISPR)144,145 and DOGMA-seq (single-cell RNA-seq, single-cell ATAC–seq and surface proteins)146. These multi-omic approaches can be used to discover new cell types147, uncover precise differentiation trajectories50,148 and characterize gene or TF regulatory networks143,144 (Fig. 3b). In the context of T cell biology, these profiling technologies have demonstrated the phenotypic diversity of intratumoral T cells50,79,82,149152, TCR repertoire evolution during development and clonal T cell dynamics after checkpoint blockade82,148,153, and can nominate new molecular regulators and therapeutic targets99,152.

Opportunities for epigenetic engineering of T cells in the clinic

Improvements in our understanding of T cell exhaustion and the mechanisms of action of currently approved immunotherapies suggest new therapeutic opportunities. In particular, the ability to modify a patient’s own T cells ex vivo and reinfuse them offers the opportunity to use engineered T cells as therapeutic agents themselves154,155 (Fig. 4a). The two most common types of T cell therapies are engineered TCR T cells, where a synthetic TCR is introduced that recognizes, for example, a known cancer antigen (that is, peptide-MHC antigen)156,157, and CAR T cells, in which a synthetic construct is introduced that combines antibody-mediated antigen recognition with an intracellular signaling domain154,158. It is increasingly recognized that exhaustion limits CAR T cell function, but can be ameliorated with various engineering strategies such as using the 4–1BB co-stimulatory domain159, overexpressing AP-1 factors, such as c-Jun or BATF83,160, knocking out NR4A family members161 or TET2 (ref. 162), and ‘resting’ the cells by limiting antigen exposure163. Historically, nearly all of these engineering efforts have relied on the use of viral delivery—lentiviral or retroviral delivery of a synthetic construct randomly into the genome—but CRISPR–Cas9 has recently enabled targeted genome engineering directly in primary T cell therapies. CRISPR–Cas9-mediated knock-in can be used to insert the antigen-recognition domain directly into the endogenous TCR locus (TRAC locus)131,164, optionally together with other genes165. CRISPR–Cas9 can also be used to inactivate specific genes in clinical-grade cell therapy products, the safety of which was recently demonstrated in a first-in-human trial166. In the coming years, we anticipate that the T cell engineering field will identify a diverse set of targets for genome engineering via high-throughput screening, and will expand to encompass more sophisticated engineering approaches such as using dual sgRNAs to knock out specific enhancers, or directly perturbing genome conformation35,127,133,165,167169 (Fig. 4b).

Fig. 4 |. Opportunities for epigenetic engineering in the clinic.

Fig. 4 |

a, Engineered T cell therapies. b, Emerging strategies for genetic and epigenetic engineering.

Future perspectives

In conclusion, although there has been marked progress in our understanding of the development of T cell exhaustion, its unique features and its importance in diverse disease settings, important questions remain. First, single-cell and longitudinal studies are needed to finely characterize the phenotypic plasticity of these cells at each step in their differentiation trajectory. For example, a recent study in humans with hepatitis C virus infection demonstrated that TEX cells that have undergone years of chronic stimulation have permanent deficits in memory formation after viral clearance110. However, T cells in which TCR signaling was stopped earlier—due to viral evolution that caused viral escape from certain TCRs but not complete viral clearance—could completely recover, consistent with findings in CAR T cells that demonstrate that antigen ‘rest’ can improve T cell function110,163. Therefore, more precise temporally resolved studies will be needed to understand exactly when TEX cells pass the ‘point of no return’ and their functional deficits become permanent, what transcriptional and epigenetic features define that state, and whether the plasticity can be extended or reversed therapeutically. These studies may benefit from recently developed lineage tracing tools, for example, using TCR sequencing to track specific T cell clones in a polyclonal setting93,170, or from recently developed tools that are compatible with transgenic TCR models that have a defined specificity171. Second, how these molecular programs coalesce with other environmental factors, such as metabolic deficiencies or stresses remains an open question172. For example, recent studies have demonstrated that hypoxia may influence the progression to terminal exhaustion36,173. Finally, high-throughput CRISPR–Cas9 screening is poised to rapidly expand our understanding of TEX cell biology and therapeutic opportunities by enabling the testing of hundreds to thousands of modifications in parallel. When performed directly in clinically relevant cell therapies such as CAR T cells, genetic ‘hits’ have a direct path to therapeutic relevance, sidestepping the labor-intensive and time-intensive search for conventional therapeutics (for example, chemical or biologic agents), which phenocopy the genetic perturbation. Furthermore, as single-cell sequencing continues to define cell states associated with efficacious cellular therapies, we envision that multimodal readouts such as Perturb-seq and Perturb-ATAC will be used to directly screen for synthetic constructs and gene knockouts that directly tune the phenotype of a particular cellular therapy toward beneficial gene expression programs, and away from dysfunctional or exhausted phenotypes. When paired with individual profiling of additional dysfunction-inducing factors such as TGF-β signaling in the TME, genetic programs tailored to a patient’s own tumor could be installed in the engineered T cell, enabling personalized cellular therapies.

Acknowledgements

We thank S. Knemeyer at SciStories for illustrations. This work was supported by the National Institutes of Health awards K08CA230188, U01CA260852 and UM1HG012076, the Parker Institute for Cancer Immunotherapy, a Career Award for Medical Scientists from the Burroughs Wellcome Fund, a Cancer Research Institute Technology Impact Award, a Baxter Foundation Faculty Scholar Award, and a Pew-Stewart Scholars for Cancer Research Award. J.A.B. was supported by a Stanford Graduate Fellowship and a National Science Foundation Graduate Research Fellowship under grant no. DGE-1656518.

Footnotes

Additional information

Peer review information Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Laurie A. Dempsey, in collaboration with the Nature Immunology team.

Competing interests

A.T.S. is a scientific founder of Immunai and founder of Cartography Biosciences and receives research funding from Merck Research Laboratories and Allogene Therapeutics. J.A.B and B.D. declare no competing interests.

REFERENCES

  • 1.Germain RN T cell development and the CD4–CD8 lineage decision. Nat. Rev. Immunol 2, 309–322 (2002). [DOI] [PubMed] [Google Scholar]
  • 2.Wherry EJ T cell exhaustion. Nat. Immunol 12, 492–499 (2011). [DOI] [PubMed] [Google Scholar]
  • 3.McLane LM, Abdel-Hakeem MS & Wherry EJ CD8+ T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol 37, 457–495 (2019). [DOI] [PubMed] [Google Scholar]
  • 4.Bengsch B. et al. Epigenomic-guided mass cytometry profiling reveals disease-specific features of exhausted CD8+ T cells. Immunity 48, 1029–1045 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Miller BC et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol 20, 326–336 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Sen DR et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165–1169 (2016). This paper used ATAC–seq to define the chromatin accessibility landscape of exhausted CD8+ T cells in LCMV clone-13 infection, which was conserved in exhausted human T cells.
  • 7. Pauken KE et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 354, 1160–1165 (2016). This paper demonstrates that, although PD-L1 blockade induces transient transcriptional reprogramming of TEX cells, the epigenetic imprint of exhaustion is stable.
  • 8.Philip M. et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 545, 452–456 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mognol GP et al. Exhaustion-associated regulatory regions in CD8+ tumor-infiltrating T cells. Proc. Natl Acad. Sci. USA 114, E2776–E2785 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Allis CD & Jenuwein T. The molecular hallmarks of epigenetic control. Nat. Rev. Genet 17, 487–500 (2016). [DOI] [PubMed] [Google Scholar]
  • 11.Andersson R. & Sandelin A. Determinants of enhancer and promoter activities of regulatory elements. Nat. Rev. Genet 21, 71–87 (2020). [DOI] [PubMed] [Google Scholar]
  • 12.Orphanides G, Lagrange T. & Reinberg D. The general transcription factors of RNA polymerase II. Genes Dev. 10, 2657–2683 (1996). [DOI] [PubMed] [Google Scholar]
  • 13.Schoenfelder S. & Fraser P. Long-range enhancer–promoter contacts in gene expression control. Nat. Rev. Genet 20, 437–455 (2019). [DOI] [PubMed] [Google Scholar]
  • 14.Hnisz D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lovén J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Whyte WA et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Vahedi G. et al. Super-enhancers delineate disease-associated regulatory nodes in T cells. Nature 520, 558–562 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kornberg RD Chromatin structure: a repeating unit of histones and DNA. Science 184, 868–871 (1974). [DOI] [PubMed] [Google Scholar]
  • 19.Olins DE & Olins AL Chromatin history: our view from the bridge. Nat. Rev. Mol. Cell Biol. 4, 809–814 (2003). [DOI] [PubMed] [Google Scholar]
  • 20.Kornberg RD & Lorch Y. Chromatin structure and transcription. Annu. Rev. Cell Biol. 8, 563–587 (1992). [DOI] [PubMed] [Google Scholar]
  • 21.Jang MK et al. The bromodomain protein Brd4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription. Mol. Cell 19, 523–534 (2005). [DOI] [PubMed] [Google Scholar]
  • 22.Yang Z. et al. Recruitment of P-TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4. Mol. Cell 19, 535–545 (2005). [DOI] [PubMed] [Google Scholar]
  • 23.Bernstein BE et al. A bivalent chromatin structure marks key developmental genes in embryonic stem. Cell 125, 315–326 (2006). [DOI] [PubMed] [Google Scholar]
  • 24.Russ BE et al. Distinct epigenetic signatures delineate transcriptional programs during virus-specific CD8+ T cell differentiation. Immunity 41, 853–865 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Solomon MJ, Larsen PL & Varshavsky A. Mapping protein–DNA interactions in vivo with formaldehyde: evidence that histone H4 is retained on a highly transcribed gene. Cell 53, 937–947 (1988). [DOI] [PubMed] [Google Scholar]
  • 26.Mikkelsen TS et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Barski A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007). [DOI] [PubMed] [Google Scholar]
  • 28.Johnson DS, Mortazavi A, Myers RM & Wold B. Genome-wide mapping of in vivo protein–DNA interactions. Science 316, 1497–1502 (2007). [DOI] [PubMed] [Google Scholar]
  • 29.Schmid M, Durussel T. & Laemmli UK ChIC and ChEC; genomic mapping of chromatin proteins. Mol. Cell 16, 147–157 (2004). [DOI] [PubMed] [Google Scholar]
  • 30.Ku WL et al. Single-cell chromatin immunocleavage sequencing to profile histone modification. Nat. Methods 16, 323–325 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kaya-Okur HS et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun 10, 1930 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Skene PJ & Henikoff S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6, e21856 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Carter B. et al. Mapping histone modifications in low cell number and single cells using antibody-guided chromatin tagmentation (ACT-seq). Nat. Commun 10, 3747 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen Y. et al. BATF regulates progenitor to cytolytic effector CD8+ T cell transition during chronic viral infection. Nat. Immunol 22, 996–1007 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Chen Z. et al. In vivo CD8+ T cell CRISPR screening reveals control by Fli1 in infection and cancer. Cell 184, 1262–1280 (2021). This paper uses in vivo screens in LCMV infection with an sgRNA pool targeting transcription factors to uncover a role for Fli1 in the effector T cell response. Fli1-deficient T cells demonstrated improved T cell immunity in multiple infection and tumor models.
  • 36.Ford BR et al. Altered co-stimulatory signals and hypoxia support chromatin landscapes limiting the functional potential of exhausted T cells in cancer. Preprint at BioRxiv 10.1101/2021.07.11.451947 (2021). [DOI] [Google Scholar]
  • 37.Wu SJ et al. Single-cell CUT&Tag analysis of chromatin modifications in differentiation and tumor progression. Nat. Biotechnol 39, 819–824 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bartosovic M, Kabbe M. & Castelo-Branco G. Single-cell CUT&Tag profiles histone modifications and transcription factors in complex tissues. Nat. Biotechnol 39, 825–835 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Luecken MD & Theis FJ Current best practices in single-cell RNA-seq analysis: a tutorial. Mol. Syst. Biol 15, e8746 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dunham I. et al. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wu C. The 5′ ends of Drosophila heat shock genes in chromatin are hypersensitive to DNase I. Nature 286, 854–860 (1980). [DOI] [PubMed] [Google Scholar]
  • 42.Boyle AP et al. High-resolution mapping and characterization of open chromatin across the genome. Cell 132, 311–322 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Schones DE et al. Dynamic regulation of nucleosome positioning in the human genome. Cell 132, 887–898 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Buenrostro JD, Giresi PG, Zaba LC, Chang HY & Greenleaf WJ Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Schep AN et al. Structured nucleosome fingerprints enable high-resolution mapping of chromatin architecture within regulatory regions. Genome Res. 25, 1757–1770 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Schep AN, Wu B, Buenrostro JD & Greenleaf WJ chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Corces MR et al. An improved ATAC–seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Jin W. et al. Genome-wide detection of DNase I hypersensitive sites in single cells and FFPE tissue samples. Nature 528, 142–146 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lu F. et al. Establishing chromatin regulatory landscape during mouse preimplantation development. Cell 165, 1375–1388 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Satpathy AT et al. Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion. Nat. Biotechnol 37, 925–936 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Greenberg MVC & Bourc’his D. The diverse roles of DNA methylation in mammalian development and disease. Nat. Rev. Mol. Cell Biol. 20, 590–607 (2019). [DOI] [PubMed] [Google Scholar]
  • 52.Harris CJ et al. A DNA methylation reader complex that enhances gene transcription. Science 362, 1182–1186 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wu X. & Zhang Y. TET-mediated active DNA demethylation: mechanism, function and beyond. Nat. Rev. Genet 18, 517–534 (2017). [DOI] [PubMed] [Google Scholar]
  • 54.Okano M, Bell DW, Haber DA & Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99, 247–257 (1999). [DOI] [PubMed] [Google Scholar]
  • 55.Frommer M. et al. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc. Natl Acad. Sci. USA 89, 1827–1831 (1992). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Meissner A. et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 454, 766–770 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lister R. et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462, 315–322 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lieberman-Aiden E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Dekker J, Rippe K, Dekker M. & Kleckner N. Capturing chromosome conformation. Science 295, 1306–1311 (2002). [DOI] [PubMed] [Google Scholar]
  • 60.Dixon JR et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Nora EP et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Davidson IF et al. DNA loop extrusion by human cohesin. Science 366, 1338–1345 (2019). [DOI] [PubMed] [Google Scholar]
  • 63.Vian L. et al. The energetics and physiological impact of cohesin extrusion. Cell 173, 1165–1178 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Fullwood MJ et al. An oestrogen-receptor-α-bound human chromatin interactome. Nature 462, 58–64 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Rao SSP et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mumbach MR et al. HiChIP: efficient and sensitive analysis of protein-directed genome architecture. Nat. Methods 13, 919–922 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mumbach MR et al. Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements. Nat. Genet 49, 1602–1612 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gennert DG et al. Dynamic chromatin regulatory landscape of human CAR T cell exhaustion. Proc. Natl Acad. Sci. USA 118, e2104758118 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Curtsinger JM & Mescher MF Inflammatory cytokines as a third signal for T cell activation. Curr. Opin. Immunol 22, 333–340 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zhang N. & Bevan MJ CD8+ T cells: foot soldiers of the immune system. Immunity 35, 161–168 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kaech SM et al. Selective expression of the interleukin 7 receptor identifies effector CD8+ T cells that give rise to long-lived memory cells. Nat. Immunol 4, 1191–1198 (2003). [DOI] [PubMed] [Google Scholar]
  • 72.Joshi NS et al. Inflammation directs memory precursor and short-lived effector CD8+ T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Youngblood B. et al. Effector CD8+ T cells dedifferentiate into long-lived memory cells. Nature 552, 404–409 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Akondy RS et al. Origin and differentiation of human memory CD8+ T cells after vaccination. Nature 552, 362–367 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Pritykin Y. et al. A unified atlas of CD8+ T cell dysfunctional states in cancer and infection. Mol. Cell 81, 2477–2493 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Day CL et al. PD-1 expression on HIV-specific T cells is associated with T cell exhaustion and disease progression. Nature 443, 350–354 (2006). [DOI] [PubMed] [Google Scholar]
  • 77.Trautmann L. et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat. Med 12, 1198–1202 (2006). [DOI] [PubMed] [Google Scholar]
  • 78.McKinney EF, Lee JC, Jayne DRW, Lyons PA & Smith KGC T cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Li H. et al. Dysfunctional CD8+ T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 176, 775–789 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Im SJ et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016). This paper identified progenitor TEX cells, demonstrated that these cells are the TEX cell subset that proliferates in response to PD-1 blockade, and showed that TCF1 is required for the formation of these cells.
  • 81.Sade-Feldman M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Yost KE et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med 25, 1251–1259 (2019). This paper used single-cell RNA and T cell receptor sequencing on patient TILs before and after anti-PD-1 therapy. The authors demonstrate that a large fraction of clonally expanded T cells after treatment is not detectable before treatment, suggesting that PD-1 blockade serves to recruit new T cell clones from the periphery.
  • 83.Lynn RC et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293–300 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Zajac AJ et al. Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med 188, 2205–2213 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Gallimore A. et al. Induction and exhaustion of lymphocytic choriomeningitis virus-specific cytotoxic T lymphocytes visualized using soluble tetrameric major histocompatibility complex class I-peptide complexes. J. Exp. Med 187, 1383–1393 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Wherry EJ et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007). [DOI] [PubMed] [Google Scholar]
  • 87.Scott-Browne JP et al. Dynamic changes in chromatin accessibility occur in CD8+ T cells responding to viral infection. Immunity 45, 1327–1340 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hudson WH et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic infection. Immunity 51, 1043–1058 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zander R. et al. CD4+ T cell help is required for the formation of a cytolytic CD8+ T cell subset that protects against chronic infection and cancer. Immunity 51, 1028–1042 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Raju S. et al. Identification of a T-bethi quiescent exhausted CD8+ T cell subpopulation that can differentiate into TIM3+CX3CR1+ effectors and memory-like cells. J. Immunol 206, 2924–2936 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Beltra J-C et al. Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 52, 825–841 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Gabriel SS et al. Transforming growth factor-β-regulated mTOR activity preserves cellular metabolism to maintain long-term T cell responses in chronic infection. Immunity 54, 1698–1714 (2021). [DOI] [PubMed] [Google Scholar]
  • 93.Daniel B. et al. Divergent clonal differentiation trajectories of T cell exhaustion. Preprint at BioRxiv 10.1101/2021.12.16.472900 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Shakiba M. et al. TCR signal strength defines distinct mechanisms of T cell dysfunction and cancer evasion. J. Exp. Med 219, e20201966 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Burger ML et al. Antigen dominance hierarchies shape TCF1+ progenitor CD8+ T cell phenotypes in tumors. Cell 184, 4996–5014 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Good CR et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell 184, 6081–6100 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Yao C. et al. BACH2 enforces the transcriptional and epigenetic programs of stem-like CD8+ T cells. Nat. Immunol 22, 370–380 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Utzschneider DT et al. Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nat. Immunol 21, 1256–1266 (2020). [DOI] [PubMed] [Google Scholar]
  • 99.Yao C. et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol 20, 890–901 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Alfei F. et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269 (2019). [DOI] [PubMed] [Google Scholar]
  • 101.Khan O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Scott AC et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Seo H. et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc. Natl Acad. Sci. USA 116, 12410–12415 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Martinez GJ et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 42, 265–278 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Barber DL et al. Restoring function in exhausted CD8+ T cells during chronic viral infection. Nature 439, 682–687 (2006). This paper reports that PD-1 is a marker of TEX cells and that antibody blockade of PD-L1 results in enhanced T cell expansion and improved viral control.
  • 106.Velu V. et al. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature 458, 206–210 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Utzschneider DT et al. T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat. Immunol 14, 603–610 (2013). [DOI] [PubMed] [Google Scholar]
  • 108.Abdel-Hakeem MS et al. Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation. Nat. Immunol 22, 1008–1019 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Yates KB et al. Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans. Nat. Immunol 22, 1020–1029 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Tonnerre P. et al. Differentiation of exhausted CD8+ T cells after termination of chronic antigen stimulation stops short of achieving functional T cell memory. Nat. Immunol 22, 1030–1041 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Ghoneim HE et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170, 142–157 (2017). This paper defines de novo DNA methylation programs acquired by TEX cells and shows that genetic or pharmacologic inhibition of methylation synergizes with anti-PD-L1 treatment to enhance T cell responses.
  • 112.Prinzing B. et al. Deleting DNMT3A in CAR T cells prevents exhaustion and enhances antitumor activity. Sci. Transl. Med 13, eabh0272 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Xia Y. et al. BCL6-dependent TCF-1+ progenitor cells maintain effector and helper CD4 T cell responses to persistent antigen. Preprint at BioRxiv 10.1101/2021.08.06.455141 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Schietinger A. et al. Tumor-specific T cell dysfunction is a dynamic antigen-driven differentiation program initiated early during tumorigenesis. Immunity 45, 389–401 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Utzschneider DT et al. High antigen levels induce an exhausted phenotype in a chronic infection without impairing T cell expansion and survival. J. Exp. Med 213, 1819–1834 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Jinek M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial. Science 337, 816–821 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Cong L. et al. Multiplex genome engineering using CRISPR–Cas systems. Science 339, 819–823 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Qi LS et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Gilbert LA et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Konermann S. et al. Genome-scale transcriptional activation by an engineered CRISPR–Cas9 complex. Nature 517, 583–588 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Wang T, Wei JJ, Sabatini DM & Lander ES Genetic screens in human cells using the CRISPR–Cas9 system. Science 343, 80–84 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Shalem O. et al. Genome-scale CRISPR–Cas9 knockout screening in human cells. Science 343, 84–87 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Gilbert LA et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Canver MC et al. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature 527, 192–197 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Fulco CP et al. Systematic mapping of functional enhancer–promoter connections with CRISPR interference. Science 354, 769–773 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Simeonov DR et al. Discovery of stimulation-responsive immune enhancers with CRISPR activation. Nature 549, 111–115 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Shifrut E. et al. Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function. Cell 175, 1958–1971 (2018). This paper demonstrates how lentiviral sgRNA pools can be used together with electroporation of Cas9 protein to enable genome-scale genetic screens in primary human T cells.
  • 128.Platt RJ et al. CRISPR–Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Parnas O. et al. A genome-wide CRISPR screen in primary immune cells to dissect regulatory networks. Cell 162, 675–686 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Schumann K. et al. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc. Natl Acad. Sci. USA 112, 10437–10442 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Roth TL et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 559, 405–409 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Henriksson J. et al. Genome-wide CRISPR screens in T helper cells reveal pervasive crosstalk between activation and differentiation. Cell 176, 882–896 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Dong MB et al. Systematic immunotherapy target discovery using genome-scale in vivo CRISPR screens in CD8+ T cells. Cell 178, 1189–1204 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Schmidt R. et al. CRISPR activation and interference screens decode stimulation responses in primary human T cells. Science 375, eabj4008 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Macosko EZ et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Han A, Glanville J, Hansmann L. & Davis MM Linking T cell receptor sequence to functional phenotype at the single-cell level. Nat. Biotechnol 32, 684–692 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Bendall SC et al. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 332, 687–696 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Stoeckius M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Replogle JM et al. Combinatorial single-cell CRISPR screens by direct guide RNA capture and targeted sequencing. Nat. Biotechnol 38, 954–961 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ma S. et al. Chromatin potential identified by shared single-cell profiling of RNA and chromatin. Cell 183, 1103–1116 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Adamson B. et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell 167, 1867–1882 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Jaitin DA et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell 167, 1883–1896 (2016). [DOI] [PubMed] [Google Scholar]
  • 143.Dixit A. et al. Perturb-seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell 167, 1853–1866 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Rubin AJ et al. Coupled single-cell CRISPR screening and epigenomic profiling reveals causal gene regulatory networks. Cell 176, 361–376 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Pierce SE, Granja JM & Greenleaf WJ High-throughput single-cell chromatin accessibility CRISPR screens enable unbiased identification of regulatory networks in cancer. Nat. Commun 12, 2969 (2021). This paper introduces a method for simultaneous profiling of single-cell ATAC–seq measurements and CRISPR perturbations in single cells, in a manner compatible with the widely used 10x Genomics commercial platform.
  • 146.Mimitou EP et al. Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nat. Biotechnol 39, 1246–1258 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Plasschaert LW et al. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 560, 377–381 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Park J-E et al. A cell atlas of human thymic development defines T cell repertoire formation. Science 367, eaay3224 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Singer M. et al. A distinct gene module for dysfunction uncoupled from activation in tumor-infiltrating T cells. Cell 166, 1500–1511 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Azizi E. et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–1308 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Jerby-Arnon L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Mathewson ND et al. Inhibitory CD161 receptor identified in glioma-infiltrating T cells by single-cell analysis. Cell 184, 1281–1298 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Wu TD et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature 579, 274–278 (2020). [DOI] [PubMed] [Google Scholar]
  • 154.Maude SL et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med 371, 1507–1517 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Lee DW et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385, 517–528 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Rapoport AP et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat. Med 21, 914–921 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Robbins PF et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T cell receptor: long-term follow-up and correlates with response. Clin. Cancer Res. 21, 1019–1027 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Fraietta JA et al. Determinants of response and resistance to CD19 chimeric antigen receptor T cell therapy of chronic lymphocytic leukemia. Nat. Med 24, 563–571 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Long AH et al. 4–1BB co-stimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med 21, 581–590 (2015). This paper identifies exhaustion as limiting CAR T cell efficacy and defines the role of the CAR co-stimulatory domain in susceptibility to exhaustion. It shows that the 4–1BB domain mitigates exhaustion while the CD28 domain exacerbates exhaustion, providing insight into the enhanced clinical efficacy observed with CAR T cells that incorporate the 4–1BB domain.
  • 160.Seo H. et al. BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells. Nat. Immunol 22, 983–995 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Chen J. et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 567, 530–534 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Fraietta JA et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 558, 307–312 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Weber EW et al. Transient rest restores functionality in exhausted CAR T cells through epigenetic remodeling. Science 372, eaba1786 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Eyquem J. et al. Targeting a CAR to the TRAC locus with CRISPR–Cas9 enhances tumour rejection. Nature 543, 113–117 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Roth TL et al. Pooled knockin targeting for genome engineering of cellular immunotherapies. Cell 181, 728–744 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Stadtmauer EA et al. CRISPR-engineered T cells in patients with refractory cancer. Science 367, eaba7365 (2020). This paper reports a first-in-human phase 1 clinical trial assessing CRISPR-modified T cell therapies. Edited T cells engrafted and persisted in human patients and no toxicities were observed, demonstrating feasibility and safety of CRISPR engineered CAR T cell therapies.
  • 167.Milner JJ et al. Runx3 programs CD8+ T cell residency in non-lymphoid tissues and tumours. Nature 552, 253–257 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Huang H. et al. In vivo CRISPR screening reveals nutrient signaling processes underpinning CD8+ T cell fate decisions. Cell 184, 1245–1261 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Wei J. et al. Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy. Nature 576, 471–476 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Pai JA & Satpathy AT High-throughput and single-cell T cell receptor sequencing technologies. Nat. Methods 18, 881–892 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Bowling S. et al. An engineered CRISPR–Cas9 mouse line for simultaneous readout of lineage histories and gene expression profiles in single cells. Cell 181, 1410–1422 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Vardhana SA et al. Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen. Nat. Immunol 21, 1022–1033 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Scharping NE et al. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat. Immunol 22, 205–215 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES